WO2012118974A2 - Détection d'expression de rétrovirus endogène humain dans des cellules cancéreuses et des cellules saines - Google Patents

Détection d'expression de rétrovirus endogène humain dans des cellules cancéreuses et des cellules saines Download PDF

Info

Publication number
WO2012118974A2
WO2012118974A2 PCT/US2012/027289 US2012027289W WO2012118974A2 WO 2012118974 A2 WO2012118974 A2 WO 2012118974A2 US 2012027289 W US2012027289 W US 2012027289W WO 2012118974 A2 WO2012118974 A2 WO 2012118974A2
Authority
WO
WIPO (PCT)
Prior art keywords
herv
antibody
nucleic acid
cancer
cell
Prior art date
Application number
PCT/US2012/027289
Other languages
English (en)
Other versions
WO2012118974A3 (fr
Inventor
Vincent K. TSIAGBE
Lloyd Old
Yu Li
Original Assignee
University Of Medicine And Dentistry Of New Jersey
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Medicine And Dentistry Of New Jersey filed Critical University Of Medicine And Dentistry Of New Jersey
Publication of WO2012118974A2 publication Critical patent/WO2012118974A2/fr
Publication of WO2012118974A3 publication Critical patent/WO2012118974A3/fr
Priority to US14/198,201 priority Critical patent/US20140256583A1/en
Priority to US15/495,627 priority patent/US10370417B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • C07K16/1045Lentiviridae, e.g. HIV, FIV, SIV
    • C07K16/1063Lentiviridae, e.g. HIV, FIV, SIV env, e.g. gp41, gp110/120, gp160, V3, PND, CD4 binding site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • C12Q1/702Specific hybridization probes for retroviruses
    • C12Q1/703Viruses associated with AIDS
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57426Specifically defined cancers leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'

Definitions

  • HERVs Human endogenous retroviruses
  • HERVs had integrated into the human genome millions of year ago, some HERV genes still have open reading frames (ORFs) and thus can potentially code for protein.
  • ORFs open reading frames
  • HERV-W expression has been detected in lung, gastric, ovarian and bladder cancers, and HERV-W has been reported to induce ⁇ 16 biased T cell response when presented as "multiple sclerosis retroviral particles.”
  • GC germinal center
  • vSAg29 retroviral superantigen
  • mtv29 endogenous mouse mammary tumor virus
  • Lymphoma-host CD4 T cell interaction was observed in which the lymphoma cells stimulated syngeneic response in PBL CD4 T cells, resulting in BV23 oligoclonality (92% of BV23 had the same CDR3 length as observed for the lymphoma- containing CD4 T cells).
  • the invention generally relates to human endogenous retrovirus env (HERV- WL) polypeptides, nucleotide sequences, HERV-WL antibodies, methods to detect cancer, and methods to determine the effectiveness of the treatment of cancer.
  • HERV- WL human endogenous retrovirus env
  • the invention provides an isolated antibody that specifically binds to a human endogenous env polypeptide (HERV-WL).
  • the antibody binds to an epitope comprising the sequence TEKVEIRDGIQRRA (SEQ ID NO:2).
  • the antibody may be a monoclonal, polyclonal, humanized, or human antibody.
  • the antibody may also be conjugated to a detectable label and/or a toxin.
  • the invention provides a method of delivering a toxin to a cell that expresses HERV-WL comprising contacting a cell with an isolated antibody that specifically binds to HERV-WL, wherein the antibody is conjugated to a toxin.
  • the invention provides a method of detecting HERV-WL in a cell comprising contacting a cell with an isolated antibody that specifically binds to HERV-WL and detecting the presence of a complex of the antibody and HERV-WL; wherein the presence of the complex is indicative of the detecting of HERV-WL in the cell.
  • the cell may be a bone cell, muscle cell, placenta cell, endothelial cell, epithelial cell, epidermoid cell, glial cell, tumor cell, or a cancer cell.
  • the detection is performed by immunoassay, ELISA, immunoprecipitation,
  • the invention provides a method of detecting a cancer in a subject comprising determining a level of HEVR-WL or a nucleic acid encoding HERV- WL in a biological sample from the subject; comparing the level detected in the subject's sample to a standard level in a control sample; and determining that a cancer is present if the level in the subject's sample is greater than the standard level in the control sample.
  • the level of HERV-WL may be detected by an antibody that specifically binds to HERV-WL.
  • the level of the nucleic acid encoding HERV-WL is determined by detecting binding of the nucleic acid to a second nucleic acid which is at least 85% identical to SEQ ID NO: l or complement thereof, or a third nucleic acid that encodes a polypeptide that is at least 85% identical to the sequence of SEQ ID NO:2 or the complement thereof.
  • the cancer may be a blood cancer, lymphoma, B cell lymphoma, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, Burkitt' s lymphoma, ameloblastoma, carcinomas including squamous cell carcinoma, mucoepidermoid carcinoma, ovarian cancer, cervical cancer, prostate cancer and breast cancer.
  • the biological sample may be peripheral blood and/or saliva.
  • the invention provides a method for determining the effectiveness of a treatment in a subject suffering from cancer comprising, obtaining a pretreatment biological sample from a subject, obtaining a post treatment biological sample from the subject, detecting the level of HERV-WL present in the samples;
  • the level of HERV-WL in the pretreatment biological sample is determined to be effective if the HERV-WL polypeptide present in the post-treatment biological sample is decreased compared to the HERV-WL level present in the pretreatment sample.
  • the level of HERV-WL may be detected by an antibody that specifically binds to HERV-WL.
  • the level of the nucleic acid encoding HERV-WL is determined by detecting binding of the nucleic acid to a second nucleic acid which is at least 85% identical to SEQ ID NO: l or complement thereof, or a third nucleic acid that encodes a polypeptide that is at least 85% identical to the sequence of SEQ ID NO:2 or the complement thereof.
  • the cancer may be a blood cancer, lymphoma, B cell lymphoma, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, Burkitt' s lymphoma, ameloblastoma, carcinomas including squamous cell carcinoma,
  • the invention provides an isolated nucleic acid which is at least 85% identical to SEQ ID NO: 1.
  • the nucleic acid hybridizes to SEQ ID NO: l under high stringency conditions.
  • the invention provides an isolated nucleic acid comprising a sequence that encodes a peptide sequence that is at least 85% identical to SEQ ID NO:2.
  • the nucleic acid hybridizes to SEQ ID NO:2 under high stringency conditions.
  • the invention provides an isolated polypeptide encoded by an isolated nucleic acid which is at least 85% identical to SEQ ID NO: 1.
  • the polypeptide is encoded by an isolated nucleic acid that hybridizes to SEQ ID NO: l under high stringency conditions.
  • the invention provides an isolated polypeptide comprising a sequence which is as least 99% identical to SEQ ID NO:2.
  • the invention provides a recombinant vector comprising an isolated nucleic acid which is at least 85% identical to SEQ ID NO: 1.
  • the recombinant vector may comprise a nucleic acid that hybridizes to SEQ ID NO: 1 under high stringency conditions.
  • the recombinant vector may comprise a nucleic acid that hybridizes to SEQ ID NO:2 under high stringency conditions.
  • the recombinant vector is a recombinant expression vector.
  • the invention provides an array that comprises a solid support having a plurality of locations and a nucleic acid which is at least 85% identical to SEQ ID NO: 1, a nucleic acid that hybridizes to SEQ ID NO: l under high stringency conditions or a nucleic acid that hybridizes to SEQ ID NO:2 under high stringency conditions, attached to the locations.
  • the invention provides a host comprising a recombinant vector that contains a nucleic acid which is at least 85% identical to SEQ ID NO: 1, a nucleic acid that hybridizes to SEQ ID NO: 1 under high stringency conditions or a nucleic acid that hybridizes to SEQ ID NO:2 under high stringency conditions.
  • the invention provides a kit for performing the methods of the invention.
  • Fig. 1 illustrates the relative expression of HERV-W mRNA (normalized to that of TBP mRNA expression) + SEM in normal tonsilar low density (“activated") B cells (40/50% percol gradient interphase, Tonsil low 8 ) as measured by Real-Time
  • Fig. 2 illustrates the relative transcription of HERV-W in normal and lymphoma B cells as measured by Real-Time Quantitative PCR.
  • FIG. 3 depicts specificity of HERV-WL detection.
  • ELISA plates were coated with equal concentrations of HERV-WL protein or control bovine serum Albumin (BSA), horse Ig, and normal rabiit Ig (NL Rblg).
  • BSA bovine serum Albumin
  • horse Ig horse Ig
  • normal rabiit Ig NL Rblg
  • Fig. 4 depicts the detection of HERV-WL protein in serum samples from normal and cancer patients.
  • Fig. 5 depicts the detection of HERV-WL protein in saliva samples from normal and cancer patients.
  • Fig. 6 is the Predicted Amino-acid translations of HERV-WL env, western blot analysis also shows two conforming major bands.
  • Fig. 6A is Frame 1 of HERV-WL env;
  • Fig. 6B is Frame 2 of HERV-WL env.
  • This invention is based, at least in part, on unexpected discoveries of a novel human endogenous retroviral env gene, HERV-WL. Novel HERV-WL polypeptides, nucleotide sequences and HERV-WL antibodies that are useful in methods to detect cancer, and methods to determine the effectiveness of the treatment of cancer.
  • HERV-WL means a novel human endogenous retroviral gene that is substantially identical and complementary to SEQ ID NO: l, and refers to all isoforms and variants of a HERV-WL polypeptide and the polynucleotide that encodes the HERV-WL polypeptide.
  • antibody refers to an immunoglobulin or antigen-binding fragment thereof, and encompasses any such polypeptide comprising an antigen-binding fragment of an antibody.
  • antibody also includes antigen-binding fragments of an antibody.
  • antigen-binding fragments include, but are not limited to, Fab fragments (consisting of the V L , V H , C L and C H I domains); Fd fragments (consisting of the V H and C H I domains); Fv fragments (referring to a dimer of one heavy and one light chain variable domain in tight, non-covalent association); dAb fragments (consisting of a V H domain); single domain fragments (V H domain, V L domain, V HH domain, or V NAR domain); isolated CDR regions; (Fab') 2 fragments, bivalent fragments (comprising two Fab fragments linked by a disulphide bridge at the hinge region), scFv (referring to a fusion of the V L and V H domains, linked together with a short linker), and other antibody fragments that retain antigen-binding function.
  • Fab fragments consisting of the V L , V H , C L and C H I domains
  • Fd fragments consististing of the V H
  • amino acid refers to natural and/or unnatural or synthetic amino acids, including glycine and both the D and L optical isomers, amino acid analogs (for example norleucine is an analog of leucine) and peptidomimetics.
  • Animal includes all vertebrate animals including humans.
  • vertebrate animal includes, but not limited to, mammals, humans, canines (e.g., dogs), felines (e.g., cats); equines (e.g., horses), bovines (e.g., cattle), porcine (e.g., pigs), mice, rabbits, goats, as well as in avians.
  • avian refers to any species or subspecies of the taxonomic class ava, such as, but not limited to, chickens (breeders, broilers and layers), turkeys, ducks, a goose, a quail, pheasants, parrots, finches, hawks, crows and ratites including ostrich, emu and cassowary.
  • Array refers to a solid support having a plurality of locations to attach a nucleotide sequence such as a probe or an antibody.
  • Attached or “immobilized' as used herein to refer to a probe or an antibody and a solid support refers to the binding between a probe or an antibody and the solid support is sufficient to be stable under conditions of binding, washing, analysis, and removal.
  • the binding may be covalent or non-covalent. Covalent bonds may be formed directly between the probe and the solid support or may be formed by a cross linker or by inclusion of a specific reactive group on either the solid support or the probe or both molecules.
  • Non-covalent binding may be one or more of electrostatic, hydrophilic, and hydrophobic interactions.
  • non-covalent binding is the covalent attachment of a molecule, such as streptavidin, to the support and the non-covalent binding of a biotinylated probe to the streptavidin. Immobilization may also involve a combination of covalent and non-covalent interactions.
  • the "solid substrate" used for the array may be in the form of beads, particles or sheets, and may be permeable or impermeable, depending on the type of array, wherein the surface is coated with a suitable material enabling binding of the binding reagents at high affinity.
  • the surface may be in the form of beads or particles, fibers (such as glass wool or other glass or plastic fibers) or glass or plastic capillary tubes.
  • the solid surface may be in the form of plastic, micromachined chips, membranes, slides, plates or sheets in which at least one surface is substantially flat, wherein these surfaces may comprise glass, plastic, silicon, low cross-linked and high cross-linked polystyrene, silica gel, polyamide, and the like.
  • Biological sample as used herein means a sample of biological tissue or fluid that comprises polypeptides and/or nucleic acids. Such samples include, but are not limited to, tissue isolated from animals. Biological samples may also include sections of tissues such as biopsy and autopsy samples, frozen sections taken for histologic purposes, blood, plasma, serum, sputum, saliva, stool, tears, mucus, hair, and skin. Biological samples also include explants and primary and/or transformed cell cultures derived from patient tissues. A biological sample may be provided by removing a sample of cells from an animal, but can also be accomplished by using previously isolated cells (e.g., isolated by another person, at another time, and/or for another purpose), or by performing the methods of the invention in vivo.
  • tissue isolated from animals include, but are not limited to, tissue isolated from animals. Biological samples may also include sections of tissues such as biopsy and autopsy samples, frozen sections taken for histologic purposes, blood, plasma, serum, sputum, saliva, stool, tears, mucus, hair, and skin.
  • biopsy and “biopsy tissue” refer to a sample of tissue or fluid that is removed from a subject for the purpose of determining if the sample contains cancerous tissue. In some embodiments, biopsy tissue or fluid is obtained because a subject is suspected of having cancer, and the biopsy tissue or fluid is then examined for the presence or absence of cancer.
  • cancer cell and “tumor cell”, and grammatical equivalents refer to the total population of cells derived from a tumor, a cancer or a pre-cancerous lesion.
  • “Complement” or “complementary” as used herein means Watson-Crick or Hoogsteen base pairing between nucleotides or nucleotide analogs of nucleic acid molecules.
  • amino acid variants refers to amino acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical or associated (e.g., naturally contiguous) sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode most proteins. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine.
  • nucleic acid variations are "silent variations", which are one species of conservatively modified variations.
  • Every nucleic acid sequence herein which encodes a polypeptide also describes silent variations of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant", including where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art.. Guidance concerning which amino acid changes are likely to be phenotypically silent can also be found in Bowie et al., 1990, Science 247: 1306 1310.
  • Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles.
  • Typical conservative substitutions include but are not limited to: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • Amino acids can be substituted based upon properties associated with side chains, for example, amino acids with polar side chains may be substituted, for example, Serine (S) and Threonine (T); amino acids based on the electrical charge of a side chains, for example, Arginine (R) and Histidine (H); and amino acids that have hydrophobic side chains, for example, Valine (V) and Leucine (L).
  • changes are typically of a minor nature, such as conservative amino acid substitutions that do not significantly affect the folding or activity of the protein.
  • humanized antibody and “engineered antibody”, as used herein, is intended to include antibodies having variable region frameworks derived from sequences of human origin. Furthermore, if the antibody contains a constant region, the constant region is typically derived from such human sequences, e.g., human germline sequences, or naturally occurring (e.g., allotypes) or mutated versions of human germline sequences.
  • the humanized antibodies of the invention may include amino acid residues not encoded by human sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • epitope refers to a site on an antigen to which B and/or T cells respond or a site on a molecule against which an antibody will be produced and/or to which an antibody will bind.
  • an epitope can be recognized by an antibody defining the epitope.
  • An epitope can be either a "linear epitope” (where a primary amino acid primary sequence comprises the epitope; typically at least 3 contiguous amino acid residues, and more usually, at least 5, and up to about 8 to about 10 amino acids in a unique sequence) or a "conformational epitope" (an epitope wherein the primary, contiguous amino acid sequence is not the sole defining component of the epitope).
  • a conformational epitope may comprise an increased number of amino acids relative to a linear epitope, as this conformational epitope recognizes a three-dimensional structure of the peptide or protein. For example, when a protein molecule folds to form a three dimensional structure, certain amino acids and/or the polypeptide backbone forming the conformational epitope become juxtaposed enabling the antibody to recognize the epitope.
  • Methods of determining conformation of epitopes include but are not limited to, for example, x-ray crystallography, two-dimensional nuclear magnetic resonance spectroscopy and site-directed spin labeling and electron paramagnetic resonance spectroscopy. See, for example, Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed. (1996), the disclosure of which is incorporated in its entirety herein by reference.
  • a "fusion polypeptide” refers to a polypeptide created through the joining of two or more heterologous proteins or polypeptides.
  • a heterologous protein, polypeptide, nucleic acid, or gene is one that originates from a foreign species, or, if from the same species, is substantially modified from its original form. Two fused domains or sequences are heterologous to each other if they are not adjacent to each other in a naturally occurring protein or nucleic acid.
  • Host cell refers to a naturally occurring cell or a transformed cell that contains a vector and supports the replication of the vector.
  • Host cells may be cultured cells, explants, cells in vivo, and the like.
  • Host cells may be prokaryotic cells such as E. coli, or eukaryotic cells such as yeast, insect, amphibian, or mammalian cells, such as CHO, HeLa.
  • nucleic acids or polypeptide sequences means that the sequences have a specified percentage of nucleotides or amino acids that are the same over a specified region. The percentage may be calculated by comparing optimally aligning the two sequences, comparing the two sequences over the specified region, determining the number of positions at which the identical residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the specified region, and multiplying the result by 100 to yield the percentage of sequence identity.
  • Label as used herein may mean a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
  • useful labels include radioactive isotopes, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens, green fluorescent protein, and other entities which can be made detectable.
  • a label may be incorporated into nucleic acids and proteins at any position.
  • linking refers to a chemical moiety that connects a molecule to another molecule, covalently links separate parts of a molecule or separate molecules.
  • the linker provides spacing between the two molecules or moieties such that they are able to function in their intended manner.
  • Examples of linking groups include peptide linkers, enzyme sensitive peptide linkers/linkers, self-immolative linkers, acid sensitive linkers, multifunctional organic linking agents, bifunctional inorganic
  • linker may be stable or degradable/cleavable.
  • nucleic acid refers to a polymer composed of a multiplicity of nucleotide units (ribonucleotide or deoxyribonucleotide or related structural variants) linked via phosphodiester bonds, including but not limited to, DNA or RNA.
  • the term encompasses sequences that include any of the known base analogs of DNA and RNA. Examples of a nucleic acid include and are not limited to mRNA, miRNA, tRNA, rRNA, snRNA, siRNA, dsRNA, cDNA and DNA/RNA hybrids.
  • Nucleic acids may be single stranded or double stranded, or may contain portions of both double stranded and single stranded sequence.
  • the nucleic acid may be DNA, both genomic and cDNA, RNA, or a hybrid, where the nucleic acid may contain combinations of deoxyribo- and ribo-nucleotides, and combinations of bases including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine hypoxanthine, isocytosine and isoguanine.
  • Nucleic acids may be obtained by chemical synthesis methods or by recombinant methods.
  • a nucleic acid also encompasses the complementary strand of a depicted single strand.
  • many variants of a nucleic acid may be used for the same purpose as a given nucleic acid.
  • a nucleic acid also encompasses substantially identical nucleic acids and complements thereof.
  • a single strand provides a probe for a probe that may hybridize to the target sequence under stringent hybridization conditions.
  • a nucleic acid also encompasses a probe that hybridizes under stringent hybridization conditions.
  • operable combination refers to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced.
  • the term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.
  • polypeptide in its broadest sense refers to a compound of two or more subunit amino acids, amino acid analogs or peptidomimetics.
  • Probe refers to an oligonucleotide capable of binding to a target nucleic acid of complementary sequence through one or more types of chemical bonds, usually through complementary base pairing, usually through hydrogen bond formation. Probes may bind target sequences lacking complete complementarity with the probe sequence depending upon the stringency of the hybridization conditions. There may be any number of base pair mismatches which will interfere with hybridization between the target sequence and the single stranded nucleic acids of the present invention.
  • a probe may be single stranded or partially single and partially double stranded. The strandedness of the probe is dictated by the structure, composition, and properties of the target sequence. Probes may be directly labeled or indirectly labeled such as with biotin to which a streptavidin complex may later bind. A probe may range in length from 5 nucleotides to a 1000 nucleotides in length, most preferably from 10 to 50 nucleotides in length.
  • Promoter refers to a synthetic or naturally-derived molecule which is capable of conferring or activating expression of a nucleic acid in a cell.
  • a promoter may comprise one or more specific regulatory elements to further enhance expression and/or to alter the spatial expression and/or temporal expression of same.
  • a promoter may also comprise distal enhancer or repressor elements, which can be located as much as several thousand base pairs from the start site of transcription.
  • a promoter may be derived from sources including viral, bacterial, fungal, plants, insects, and animals.
  • a promoter may regulate the expression of a gene component constitutively, or differentially with respect to cell, the tissue or organ in which expression occurs or, with respect to the developmental stage at which expression occurs, or in response to external stimuli such as physiological stresses, pathogens, metal ions, or inducing agents.
  • promoters include the bacteriophage T7 promoter, bacteriophage T3 promoter, SP6 promoter, lac operator-promoter, tac promoter, SV40 late promoter, SV40 early promoter, RSV-LTR promoter, CMV IE promoter, SV40 early promoter or SV40 late promoter and the CMV IE promoter.
  • a "recombinant" peptide, polypeptide, or protein refers to a peptide, polypeptide, or protein produced by recombinant DNA techniques; i.e., produced from cells transformed by an exogenous DNA construct encoding the desired peptide, polypeptide, or protein.
  • standard level refers to a control level of a particular protein expressed in samples of the same type of tissue or cells from subjects who do not have cancer; for example, a predetermined standard can be a control level determined based upon the expression of the HERV-WL gene in tissue isolated from subjects who do not have breast cancer.
  • Selectable marker refers to any gene which confers a phenotype on a cell in which it is expressed to facilitate the identification and/or selection of cells which are transfected or transformed with a genetic construct.
  • selectable markers include the ampicillin-resistance gene, tetracycline- resistance gene, bacterial kanamycin-resistance gene, zeocin resistance gene, the AURI-C gene which confers resistance to the antibiotic aureobasidin A, phosphinothricin-resistance gene, neomycin phosphotransferase gene (nptll), hygromycin-resistance gene, beta- glucuronidase (GUS) gene, chloramphenicol acetyltransferase (CAT) gene, green fluorescent protein-encoding gene and luciferase gene.
  • Stringent hybridization conditions refers to conditions under which a first nucleic acid sequence (e.g., probe) will hybridize to a second nucleic acid sequence (e.g., target), such as in a complex mixture of nucleic acids, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. One with ordinary skill can determine the appropriate conditions according to standard assays known in the art.
  • the term “subject” refers to any animal (e.g., a mammal), including, but not limited to humans, non-human primates, rodents, and the like, which is to be the recipient of a particular treatment.
  • the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.
  • substantially complementary refers to that a first sequence is at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical to the complement of a second sequence over a region of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more nucleotides, or that the two sequences hybridize under stringent hybridization conditions.
  • substantially identical refers to that a first and second sequence are at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identical over a region of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more nucleotides or amino acids, or with respect to nucleic acids, if the first sequence is substantially complementary to the complement of the second sequence.
  • tumor refers to any mass of tissue that results from excessive cell growth or proliferation, either benign (noncancerous) or malignant (cancerous), including pre-cancerous lesions.
  • Vector refers to a nucleic acid sequence containing an origin of replication.
  • a vector may be a plasmid, bacteriophage, bacterial artificial chromosome, yeast artificial chromosome or a virus.
  • a vector may be a DNA or RNA vector.
  • a vector may be either a self-replicating extrachromosomal vector or a vector which integrates into a host genome.
  • expression vector refers to a nucleic acid assembly containing a promoter which is capable of directing the expression of a sequence or gene of interest in a cell. Vectors typically contain nucleic acid sequences encoding selectable markers for selection of cells that have been transfected by the vector.
  • vector construct refers to any nucleic acid construct capable of directing the expression of a gene of interest and which can transfer gene sequences to target cells or host cells.
  • the present invention provides isolated antibodies that specifically bind to the human endogenous retrovirus env polypeptide (HERV-WL).
  • SEQ ID NO: l is a nucleotide sequence that encodes the HERV-WL polypeptide.
  • the antibody of the invention which is against HERV-WL, can be obtained by methods known in the art, for example by immunizing an animal with HERV-WL or an polypeptide selected from the amino acid sequence of HERV-WL to which an antibody of the invention specifically binds with measurable affinity, or an epitope selected within the HERV-WL polypeptide, and collecting and purifying the antibody produced in vivo according to a common procedure, known to those with skill in the art.
  • the HERV-WL polypeptide contains a peptide region TEKVKEIRDGIQRRA (SEQ ID NO:2).
  • Routine methods to produce HERV-WL polypeptide a polypeptide selected from the amino acid sequence of HERV-WL to which an antibody of the invention specifically binds with measurable affinity, or a polypeptide of SEQ ID NO:2 as an epitope on an antigen are known in the art.
  • HERV-WL polypeptide a polypeptide selected from the amino acid sequence of HERV-WL to which an antibody of the invention specifically binds with measurable affinity, or a polypeptide of SEQ ID NO:2 can be used as an epitope on an antigen and an animal can be immunized with HERV-WL polypeptide, apolypeptide selected from the amino acid sequence of HERV-WL to which an antibody of the invention specifically binds with measurable affinity, or SEQ ID NO:2.
  • the antigen may comprise an epitope that is substantially identical to HERV-WL polypeptide, a polypeptide selected from the amino acid sequence of HERV-WL to which an antibody of the invention specifically binds with measurable affinity, or a polypeptide of SEQ ID NO:2.
  • the epitope may also be a conservatively modified variant of HERV-WL polypeptide, a polypeptide selected from the amino acid sequence of HERV-WL to which an antibody of the invention specifically binds with measurable affinity, or a polypeptide of SEQ ID NO:2.
  • a monoclonal antibody can be obtained by fusing antibody-producing cells which produce an antibody against HERV-WL with myeloma cells to establish a hybridoma according to known methods (for example, Kohler and Milstein, Nature, (1975) 256, pp. 495-497; Kennet, R. ed., Monoclonal Antibodies, pp. 365-367, Plenum Press, N.Y. (1980)).
  • the antibody of this invention also includes humanized HERV-WL antibodies.
  • the use of transgenic mice carrying human immunoglobulin (Ig) loci in their germline configuration provide for the isolation of high affinity fully human monoclonal antibodies directed against a variety of targets including human self antigens for which the normal human immune system is tolerant (see for example Lonberg, N. et al., U.S. Pat. NO: 5,569,825, U.S. Pat. NO: 6,300,129; Kucherlapati, et al., U.S. Pat. NO: 6,713,610).
  • the endogenous immunoglobulin loci in such mice can be disrupted or deleted to eliminate the capacity of the animal to produce antibodies encoded by endogenous genes.
  • Antibodies obtained from non-human sources can be humanized.
  • the humanized antibody can comprise portions derived from an immunoglobulin of nonhuman origin with the requisite specificity, such as a mouse, and from immunoglobulin sequences of human origin (e.g., chimeric immunoglobulin), joined together chemically by conventional techniques (e.g., synthetic) or prepared as a contiguous polypeptide using genetic engineering techniques (e.g., DNA encoding the protein portions of the chimeric antibody can be expressed to produce a contiguous polypeptide chain).
  • a humanized immunoglobulin is one containing one or more immunoglobulin chains comprising a CDR derived from an antibody of nonhuman origin and a framework region derived from a light and/or heavy chain of human origin (e.g., CDR-grafted antibodies with or without framework changes).
  • the framework adaptation process was based upon the similarity of framework regions between mouse mAb C836 and sequences in the human germline databases as essentially described in WO/08052108 A2 "Methods For Use In Human- Adapting Monoclonal Antibodies" where framework length is matched residue for residue to the parental variable or V-regions.
  • Antibodies that bind to HERV-WL or an epitope selected within the HERV- WL polypeptide can be obtained using phage display technology.
  • antibodies that bind to HERV-WL polypeptide, an arbitrary polypeptide selected from the amino acid sequence of HERV-WL, and SEQ ID NO:2 can also be obtained using phage display technology. Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman et al., J. Immunol.
  • the present invention also includes nucleotide sequences that encode SEQ ID NO:2 and vectors that contain the nucleotide sequences that encode SEQ ID NO:2.
  • a human HERV-WL antibody is selected from a phage library, where that phage comprises human immunoglobulin genes and the library expresses human antibody binding domains as, for example, single chain antibodies (scFv), as Fab, or some other construct exhibiting paired or unpaired antibody variable regions fused to one or more of the phage coat proteins.
  • scFv single chain antibodies
  • Fab fragment antigen binding domains
  • Such phage display methods for isolating human antibodies are established in the art, see for example: U.S. Pat. Nos. 5,223,409; 5,403,484; and 5,571,698 to Ladner et al.; U.S. Pat. Nos.
  • the antibodies that bind to HERV-WL and SEQ ID NO:2 may also be detectably labeled.
  • labels include radioactive isotopes, enzymes, enzyme fragments, enzyme substrates, enzyme inhibitors, coenzymes, catalysts, fluorophores, dyes, chemiluminescers, luminescers, or sensitizers; a non-magnetic or magnetic particle, iodinated sugars that are used as radioopaque agents, and can be appended to a linker and conjugated to the antibody.
  • agents may be conjugated to the antibodies of this invention with or without a linker, including a toxin and/or a therapeutic agent such as small molecules that are known in the art to treat cancer, for example including without being limited, monomethyl auristatin E (MMAE), capecitabine, daunorubicin, doxorubicin, methotrexate, and Mitomycin C.
  • MMAE monomethyl auristatin E
  • An "immunotoxin” is a hybrid molecule, created by conjugating an antibody to all or part of a toxin, generally the active component of the toxin.
  • the present invention provides an immunotoxin, an HERV-WL antibody conjugated to a toxin.
  • toxins include without being limited, plant toxins such as arericin, saporin, and pokeweed antiviral protein (PAP), which inactivate ribosomes; and single-chain bacterial toxins such as diphtheria toxin (DT) and Pseudomonas exotoxin (PE). More than one agent may be conjugated to the antibody.
  • plant toxins such as arericin, saporin, and pokeweed antiviral protein (PAP), which inactivate ribosomes
  • PAP pokeweed antiviral protein
  • single-chain bacterial toxins such as diphtheria toxin (DT) and Pseudomonas exotoxin (PE).
  • DT diphtheria toxin
  • PE Pseudomonas exotoxin
  • More than one agent may be conjugated to the antibody.
  • BIOCONJUGATE TECHNIQUES Academic Press; 1st edition, Greg T. Hermanson, 1996) describes techniques for modifying or crosslinking
  • This invention also provides methods to detect HERV-WL in a cell, methods to detecting cancer in a subject and methods to assess effectiveness of a treatment in a subject suffering from cancer, as well as monitor the treatment of the cancer.
  • a subject having cancer or prone to it can be determined based on the expression levels, patterns, or profile of the HERV-WL gene, such as nucleic acids (e.g., mRNA, miRNA) or polypeptides in a biological sample from the subject compared to a standard level in a corresponding control sample, such as a non-cancerous sample.
  • HERV- WL polypeptides and nucleic acids can be used as markers to indicate the presence or absence of cancer or the risk of having cancer, as well as to assess the prognosis of the cancer.
  • Diagnostic and prognostic assays of the invention include methods for assessing the expression level of the HERV-WL nucleic acids or polypeptides.
  • An HERV-WL antibody can be used to detect the HERV-WL polypeptides.
  • a nucleotide sequence that is substantially complementary, and/or substantially identical to HERV-WL SEQ ID NO: l can be used to detect the expression level of HERV-WL.
  • a nucleotide sequence that is at least 85% identical to SEQ ID NO: l can be used to detect the expression level of HERV- WL.
  • a nucleotide sequence that is at least 99% identical to the nucleotide sequence encoded by SEQ ID NO:2 can be used to detect the expression level of HERV-WL.
  • probes that are substantially identical or substantially complementary to SEQ ID NO: 1 can be used to identify HERV-WL, as well as probes that hybridize to HERV-WL under high stringency conditions. The methods and kits herein described allow one to detect the HERV-WL in a cell, the presence of cancer in a subject and monitor the effectiveness of a cancer treatment.
  • An increased expression or presence of HERV-WL in a biological sample from a subject compared to a standard level in a control sample may be indicative that the subject has cancer. Also a relative decrease in the expression level of HERV-WL may be indicative of the decrease in the size of a tumor or growth of a cancer, further facilitating a clinician to determine whether a cancer treatment is effective.
  • HERV-WL can be detected in a variety of cell types including without limitation producer cells including bone cells, muscle cells, placenta cells, endothelial cells, epithelial cells, epidermoid cells, glial cells, tumor cells, and cells derived from tumor cell lines, and cancer cells.
  • producer cells including bone cells, muscle cells, placenta cells, endothelial cells, epithelial cells, epidermoid cells, glial cells, tumor cells, and cells derived from tumor cell lines, and cancer cells.
  • the types of cells derived from cancers such as blood cancers, lymphoma, B cell lymphoma, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, Burkitt's lymphoma, ameloblastoma, carcinomas including squamous cell carcinoma, mucoepidermoid carcinoma, ovarian cancer, cervical cancer, prostate cancer, and breast cancer.
  • the presence, level, or absence of the HERV-WL nucleic acid or polypeptide in a biological sample can be evaluated by obtaining a biological sample from a subject and contacting the biological sample with a compound or an agent capable of detecting the HERV-WL polypeptide or HERV-WL nucleic acid (e.g., mRNA, miRNA or genomic DNA probe).
  • a compound or an agent capable of detecting the HERV-WL polypeptide or HERV-WL nucleic acid e.g., mRNA, miRNA or genomic DNA probe.
  • the level of expression of HERV-WL can be measured in a number of ways, including measuring the HERV-WL mRNA; or measuring the amount of HERV- WL polypeptide.
  • RNA samples can be isolated from biological samples using any of a number of well-known procedures.
  • biological samples can be lysed in a guanidinium-based lysis buffer, optionally containing additional components to stabilize the RNA.
  • the lysis buffer can contain purified RNAs as controls to monitor recovery and stability of RNA from cell cultures. Examples of such purified RNA templates include the Kanamycin Positive Control RNA from PROMEGA (Madison, WI), and 7.5 kb Poly(A)-Tailed RNA from LIFE TECHNOLOGIES (Rockville, MD). Lysates may be used immediately or stored frozen at, e.g., -80°C.
  • total RNA can be purified from cell lysates (or other types of samples) using silica-based isolation in an automation-compatible, 96-well format, such as the RNEASY purification platform (QIAGEN, Inc., Valencia, CA).
  • RNA is isolated using solid-phase oligo-dT capture using oligo-dT bound to microbeads or cellulose columns. This method has the added advantage of isolating mRNA from genomic DNA and total RNA, and allowing transfer of the mRNA-capture medium directly into the reverse transcriptase reaction.
  • Other RNA isolation methods are contemplated, such as extraction with silica-coated beads or guanidinium. Further methods for RNA isolation and preparation can be devised by one skilled in the art.
  • RNAse inhibitors are optionally added to the crude samples.
  • genomic DNA can contribute one or more copies of a target sequence, depending on the sample. In situations in which the target sequence is derived from one or more highly expressed genes, the signal arising from genomic DNA may not be significant. But for genes expressed at low levels, the background can be eliminated by treating the samples with DNAse, or by using primers that target splice junctions for subsequent priming of cDNA or amplification products. For example, one of the two target- specific primers could be designed to span a splice junction, thus excluding DNA as a template.
  • the two target-specific primers can be designed to flank a splice junction, generating larger PCR products for DNA or unspliced mRNA templates as compared to processed mRNA templates.
  • One skilled in the art could design a variety of specialized priming applications that would facilitate use of crude extracts as samples for the purposes of this invention.
  • the level of mRNA corresponding to HERV-WL in a cell can be determined both in situ and in vitro.
  • Messenger RNA isolated from a biological sample can be used in hybridization or amplification assays that include, Southern or Northern analyses, PCR analyses, and probe arrays.
  • a preferred diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid probe that can hybridize to the mRNA encoded by HERV-WL.
  • the HERV-WL probe can be a full-length nucleic acid, for example SEQ ID NO: 1 or a portion thereof, for example the nucleotide sequence encoded by SEQ ID NO 2, an oligonucleotide of at least 10 nucleotides in length and sufficient to specifically hybridize under stringent conditions to the mRNA.
  • mRNA (or cDNA prepared from it) is immobilized on a surface and contacted with the probes, for example, by running the isolated mRNA on an agarose gel and transferring the mRNA from the gel to a membrane, such as nitrocellulose.
  • the probes are immobilized on a solid support and the mRNA (or cDNA) is contacted with the probes, for example, in a polynucleotide chip array.
  • a skilled artisan can adapt known mRNA detection methods for detecting the level of an mRNA.
  • the level of mRNA (or cDNA prepared from it) in a sample encoded by HERV-WL to be examined can be evaluated with nucleic acid amplification, e.g., by standard PCR (U.S. Patent NO: 4,683,202), RT-PCR (Bustin S. J Mol Endocrinol. 25: 169- 93, 2000), quantitative PCR (Ong Y. et al., Hematology. 7:59-67, 2002), real time PCR (Ginzinger D. Exp Hematol. 30:503-12, 2002), and in situ PCR (Thaker V. Methods Mol Biol. 115:379-402, 1999), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques known in the art.
  • primer refers to any nucleic acid that is capable of hybridizing at its 3' end to a complementary nucleic acid molecule, and that provides a free 3' hydroxyl terminus which can be extended by a nucleic acid polymerase.
  • amplification primers are a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule having the nucleotide sequence flanked by the primers.
  • a cell or tissue sample can be prepared and immobilized on a support, such as a glass slide, and then contacted with a probe that can hybridize to mRNA.
  • a probe that can hybridize to mRNA.
  • Alternative methods for amplifying nucleic acids corresponding to expressed RNA samples include those described in, e.g., U.S. Patent NO: 7,897,750.
  • the methods of the invention further include contacting a control sample with a compound or agent capable of detecting the mRNA of HERV-WL and comparing the presence of the mRNA in the control sample with the presence of the RNA in the biological sample.
  • the above-described nucleic acid-based diagnostic methods can provide qualitative and quantitative information to determine whether a subject has a disease characterized by cancer or the effectiveness of a cancer treatment.
  • An increased expression or presence of HERV-WL in a biological sample from a subject compared to a standard level in a control sample may be indicative that the subject has cancer.
  • a relative decrease in the expression level of HERV-WL may be indicative of the decrease in the size of a tumor or growth of a cancer.
  • the types of cancer include without limitation blood cancers, lymphoma, B cell lymphoma, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, Burkitt's lymphoma, carcinomas, including squamous cell carcinoma and mucoepidermoid carcinoma, cervical cancer, prostate cancer,, ovarian cancer and breast cancer.
  • a variety of methods can be used to determine the level of the HERV-WL polypeptide disclosed herein. In general, these methods include contacting an agent that selectively binds to the polypeptide, such as an antibody, to evaluate the level of polypeptide in a sample.
  • Antibodies can be polyclonal, or monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can also be used. In a preferred embodiment, the antibody bears a detectable label.
  • a label may be incorporated into the polypeptide at any position.
  • the term "labeled”, with regard to the antibody, is intended to encompass direct labeling of the probe or antibody by physically linking a detectable substance to the probe or antibody, as well as indirect labeling of the antibody by reactivity with a detectable substance.
  • an antibody with a rabbit Fc region can be indirectly labeled using a second antibody directed against the rabbit Fc region, wherein the second antibody is coupled to a detectable substance.
  • detectable substances include radio isotopes (e.g., 1251, 1311, 35S, 3H, or 32P), enzymes (e.g., alkaline phosphatase, horseradish peroxidase, luciferase, or ⁇ -glactosidase), fluorescent moieties or proteins (e.g., fluorescein, rhodamine, phycoerythrin, Green Flourescent Protein (GFP), or Blue Fluorescent Protein (BFP)), or luminescent moieties (e.g., QdotTM nanoparticles by the Quantum Dot Corporation, Palo Alto, CA).
  • radio isotopes e.g., 1251, 1311, 35S, 3H, or 32P
  • enzymes e.g., alkaline phosphat
  • the detection methods can be used to detect a polypeptide in a biological sample in vitro as well as in vivo.
  • In vitro techniques for detection of the polypeptide include ELISAs, immunoprecipitations, immunofluorescence, immunohistochemistry; immunocytochemistry; flow cytometry; antibody arrays and Western blotting analysis. Such techniques are commonly known in the art.
  • In vivo techniques for detection of the polypeptide include introducing into a subject a labeled HERV-WL antibody.
  • the antibody can be labeled with a detectable substance as described above. The presence and location of the detectable substance in a subject can be detected by standard imaging techniques.
  • An ELISA short for Enzyme-Linked Immunosorbent Assay, is a biochemical technique to detect the presence of an antibody or an antigen in a sample. It utilizes a minimum of two antibodies, one of which is specific to the antigen and the other of which is coupled to an enzyme. The second antibody will cause a chromogenic or fluorogenic substrate to produce a signal. Variations of ELISA include sandwich ELISA, competitive ELISA, and ELISPOT. Because the ELISA can be performed to evaluate either the presence of antigen or the presence of antibody in a sample, it is a useful tool both for determining serum antibody concentrations and also for detecting the presence of antigen.
  • Immunohistochemistry and immunocytochemistry refer to the process of localizing proteins in a tissue section or cell, respectively, via the principle of antigens in tissue or cells binding to their respective antibodies. Visualization is enabled by tagging the antibody with color producing or fluorescent tags. Typical examples of color tags include, but are not limited to, horseradish peroxidase and alkaline phosphatase. Typical examples of fluorophore tags include, but are not limited to, fluorescein isothiocyanate (FITC) or phycoerythrin (PE).
  • FITC fluorescein isothiocyanate
  • PE phycoerythrin
  • Flow cytometry is a technique for counting, examining and sorting microscopic particles suspended in a stream of fluid. It allows simultaneous multiparametric analysis of the physical and/or chemical characteristics of single cells flowing through an optical/electronic detection apparatus.
  • a beam of light e.g., a laser
  • a number of detectors are aimed at the point where the stream passes through the light beam; one in line with the light beam (Forward Scatter or FSC) and several perpendicular to it (SSC) and one or more fluorescent detectors).
  • Each suspended particle passing through the beam scatters the light in some way, and fluorescent chemicals in the particle may be excited into emitting light at a lower frequency than the light source.
  • the combination of scattered and fluorescent light is picked up by the detectors, and by analyzing fluctuations in brightness at each detector, one for each fluorescent emission peak, it is possible to deduce various facts about the physical and chemical structure of each individual particle.
  • FSC correlates with the cell volume and SSC correlates with the density or inner complexity of the particle (e.g., shape of the nucleus, the amount and type of cytoplasmic granules or the membrane roughness).
  • the diagnostic methods described herein to detect HERV-WL polypeptides in a biological sample from a subject can identify subjects having, or at risk of developing cancer, or the effectiveness of cancer treatment.
  • An increased expression or presence of HERV-WL in a biological sample from a subject compared to a standard level in a control sample may be indicative that the subject has cancer.
  • a relative decrease in the expression level of HERV-WL may be indicative of the decrease in the size of a tumor or growth of a cancer.
  • the types of cancer include without limitation blood cancers, lymphoma, B cell lymphoma, non-Hodgkin's lymphoma, diffuse large B cell lymphoma, Burkitt's lymphoma, carcinomas including squamous cell carcinoma, mucoepidermoid carcinoma, ovarian cancer, cervical cancer, prostate cancer, and breast cancer.
  • the present invention provides an isolated nucleic acid comprising a nucleotide sequence referred to in SEQ ID NO: l and the nucleotide sequence that encodes SEQ ID NO: 2 or variants thereof.
  • the variant may be a complement of the referenced nucleotide sequence.
  • the variant may also be a nucleotide sequence that is substantially identical to the referenced nucleotide sequence or the complement thereof.
  • the variant may also be a nucleotide sequence which hybridizes under stringent conditions to the referenced nucleotide sequence, complements thereof, or nucleotide sequences substantially identical thereto.
  • the nucleic acid may have a length of from 10 to 1700 nucleotides.
  • the nucleic acid may have a length of at least 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 60, 70, 80 or 90 nucleotides.
  • the nucleic acid may be synthesized or expressed in a cell (in vitro or in vivo) using a vector described below.
  • the nucleic acid may be synthesized as a single strand molecule and hybridized under stringent hybridization conditions to a substantially complementary nucleic acid to form a duplex, which is considered a nucleic acid of the invention.
  • the nucleic acid may be introduced to a cell, tissue or organ in a single- or double- stranded form or capable of being expressed by a vector using methods well known to those skilled in the art.
  • MOLECULAR CLONING A LABORATORY MANUAL (3rd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 2001) describes commonly used techniques in molecular biology.
  • nucleic acid for example deletions, insertions, and/or substitutions in the sequence, which cause for so-called “silent changes", are considered to be part of the invention.
  • nucleic acids of the present invention comprise also such nucleic acid sequences which contain sequences in essence equivalent to the nucleic acids described in SEQ ID NOs. 1 and 2.
  • nucleic acids can show for example at least about 70%, more typically at least about 85%, 90% or 95% sequence identity to the nucleic acid sequences of SEQ ID NOs. 1 and a nucleotide sequence that encodes the polypeptide of SEQ ID NO:2.
  • the present invention provides a vector comprising a nucleic acid comprising a sequence which is at least 85% identical to SEQ ID NO: l.
  • the present invention also provides a vector comprising a nucleic acid comprising a sequence that encodes a peptide sequence that is at least 85% identical to SEQ ID NO:2.
  • the present invention also provides a vector comprising a nucleic acid which hybridizes to the nucleic acid of SEQ ID NO: l and or a nucleotide sequence that encodes the polypeptide of SEQ ID NO:2 under high stringency conditions.
  • An expression vector may comprise additional elements.
  • the expression vector may have two replication systems allowing it to be maintained in two organisms, e.g., in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification.
  • the expression vector may contain at least one sequence homologous to the host cell genome, and preferably two homologous sequences which flank the expression construct.
  • the integrating vector may be directed to a specific locus in the host cell by selecting the appropriate homologous sequence for inclusion in the vector.
  • the vector may also comprise a selectable marker to allow the selection of transformed host cells.
  • the present invention provides a host cell comprising a vector of the invention.
  • the cell may be a bacterial, fungal, plant, insect or animal cell.
  • Methods are known in the art to introduce the nucleic acids of the present invention to a host cell, including electroporation and standard methods of transfection.
  • MOLECULAR CLONING A LABORATORY MANUAL (3rd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 2001) describes commonly used techniques in molecular biology.
  • the present invention provides an isolated polypeptide comprising a sequence encoded by SEQ ID NO: l, or having the sequence of SEQ ID NO:2 or variants thereof.
  • polypeptides can show for example at least about 70%, more typically at least about 85%, 90% or 95% sequence identity to the polypeptide sequences described in SEQ ID NOs. 1 and 2.
  • the variants of the invention can be operatively linked to a polypeptide not according to the invention (e.g., heterologous amino acid sequences) to form fusion proteins.
  • a polypeptide not according to the invention in this context refers to a polypeptide having an amino acid sequence corresponding to a polypeptide which is not substantially identical to a variant HERV-WL of the invention.
  • the variant of the invention can correspond to a full length sequence.
  • the polypeptide not according to the invention can be fused to the N- terminus or C-terminus of the variant polypeptide.
  • the fusion protein is a fusion protein in which the variant sequence/s is/are fused to the C- terminus of the GST sequences.
  • Such fusion proteins can facilitate the purification of a recombinant variant according to the invention.
  • the fusion protein is a variant of the invention containing a heterologous signal sequence at its N-terminus.
  • expression and/or secretion of a variant of the invention can be increased through use of a heterologous signal sequence.
  • the gp67 secretory sequence of the baculovirus envelope protein can be used as a heterologous signal sequence (Current Protocols in Molecular Biology, Ausubel et al., eds., John Wiley & Sons, 1992).
  • Other examples of eukaryotic heterologous signal sequences include the secretory sequences of melittin and human placental alkaline phosphatase (Stratagene; La Jolla, Calif.).
  • useful prokarytic heterologous signal sequences include the phoA secretory signal (Sambrook et al., supra) and the protein A secretory signal (Pharmacia Biotech; Piscataway, N.J.).
  • a signal sequence can be used to facilitate secretion and isolation of a variant of the invention.
  • Signal sequences are typically characterized by a core of hydrophobic amino acids, which are generally cleaved from the mature protein during secretion in one or more cleavage events.
  • Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway.
  • the signal sequence may direct secretion of the variant, such as from a eukaryotic host into which the expression vector is transformed, and the signal sequence may then be subsequently or concurrently cleaved.
  • the variant of the invention may then be readily purified from the extracellular medium by known methods.
  • the signal sequence can be linked to the variant of interest using a sequence, which facilitates purification, such as with a GST domain.
  • the sequence encoding the variant of the invention may be fused to a marker sequence, such as a sequence encoding a peptide, which facilitates purification of the fused variant of the invention.
  • the marker sequence is a hexa- histidine peptide, such as the tag provided in a pQE vector (Qiagen, Inc.), among others, many of which are commercially available. As described in Gentz et al, Proc. Natl. Acad. Sci.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • the HA tag is another peptide useful for purification which corresponds to an epitope derived of influenza hemaglutinin protein, which has been described by Wilson et al., Cell37:767 (1984), for instance.
  • the present invention provides a kit comprising one or more detection reagents which specifically bind to a HERV-WL polypeptide.
  • the kit includes an antibody that binds to the polypeptide encoded by SEQ ID NO: l or the polypeptide of SEQ ID NO:2.
  • the detection reagents may be peptide sequences known to flank the HERV-WL antibodies which bind to the HERV-WL polypeptides.
  • the reagents may be bound to a solid matrix or packaged with reagents for binding them to the matrix.
  • the solid matrix or substrate may be in the form of beads, plates, tubes, dip sticks, strips or biochips. Biochips or plates with addressable locations and discreet microtitre plates are particularly useful.
  • Detection reagents include wash reagents and reagents capable of detecting bound antibodies (such as labeled secondary antibodies), or reagents capable of reacting with the labeled antibody.
  • the kit will also conveniently include a control reagent (positive and/or negative) and/or a means for detecting the antibody. Instructions for use may also be included with the kit. Most usually, the kits will be formatted for assays known in the art, for example, ELISA assays, as are known in the art.
  • the kit may be comprised of one or more containers and may also include collection equipment, for example, bottles, bags (such as intravenous fluids bags), vials, syringes, and test tubes. Other components may include needles, diluents and buffers.
  • the kit may include at least one container comprising a pharmaceutically- acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • the kit may be customized for use in various antibody binding assays, such as competitive binding assays, non-competitive assays, direct and indirect sandwich assays, ELISAs, fluoroimmunoassays, immunoradiometric assays, luminescence assays, chemiluminesence assays, enzyme linked immunofluorescent assays (ELIFA), immunoprecipitation assays, immunohistochemistry; immunocytochemistry; and flow cytometry.
  • ELIFA enzyme linked immunofluorescent assays
  • the present invention provides nucleotide sequences to identify nucleotide sequences that encode the HERV-WL polypeptide in a biological sample.
  • Such nucleotide sequences include SEQ ID NO: l and a nucleotide sequence encoding the polypeptide of SEQ ID NO:2, and fragments thereof, and the complement of SEQ ID NO: l and a nucleotide sequence encoding the polypeptide of SEQ ID NO:2, and fragments thereof, and sequences that hybridize to the foregoing sequences under high stringency conditions, and the primers disclosed herein.
  • One with ordinary skill in the art using common recombinant methods can determine the nucleotide sequences to be used to detect a nucleotide that encodes the HERV-WL polypeptide.
  • kits to determine the presence of HERV-WL in a biological sample.
  • a kit may contain a nucleic acid described herein together with any or all of the following: assay reagents, buffers, probes and/or primers, and sterile saline or another pharmaceutically acceptable emulsion and suspension base.
  • the kit may include instructional materials containing directions (e.g., protocols) for the practice of the methods described herein.
  • the kit may be a kit for the amplification, detection, identification or quantification of a target HERV-WL mRNA sequence.
  • the kit may contain a poly(T) primer, a forward primer, a reverse primer, and a probe.
  • a kit of the invention includes one or more microarray slides (or alternative microarray format) onto which a plurality of different nucleic acids (each corresponding to different regions or nucleic acid variants of HERV-WL) have been deposited.
  • the kit can also include a plurality of labeled probes.
  • the kit can include a plurality of nucleotide sequences suitable as probes and a selection of labels suitable for customizing the included nucleotide sequences, or other nucleotide sequences at the discretion of the practitioner.
  • at least one included nucleotide sequence corresponds to a control sequence, e.g., a "housekeeping" gene, ⁇ -actin or the like.
  • Exemplary labels include, but are not limited to, a fluorophore, a dye, a radiolabel, an enzyme tag, that is linked to a nucleic acid primer.
  • kits that are suitable for amplifying nucleic acid corresponding to the expressed RNA samples are provided.
  • a kit includes reagents and primers suitable for use in any of the amplification methods described above.
  • the kits are suitable for amplifying a signal corresponding to hybridization between a probe and a target nucleic acid sample (e.g., deposited on a microarray).
  • kits one or more materials and/or reagents required for preparing a biological sample for HERV-WL expression analysis are optionally included in the kit.
  • one or more enzymes suitable for amplifying nucleic acids including various polymerases (RT, Taq, etc.), one or more deoxynucleotides, and buffers to provide the necessary reaction mixture for amplification.
  • kits are employed for analyzing HERV-WL expression patterns using mRNA as the starting template.
  • the mRNA template may be presented as either total cellular RNA or isolated mRNA; both types of sample yield comparable results.
  • the methods and kits described in the present invention allow quantitation of other products of gene expression, including tRNA, rRNA, or other transcription products.
  • kits of the invention further include software to expedite the generation, analysis and/or storage of data, and to facilitate access to databases.
  • the software includes logical instructions, instructions sets, or suitable computer programs that can be used in the collection, storage and/or analysis of the data. Comparative and relational analysis of the data is possible using the software provided.
  • cDNA was synthesized using Superscript II reagents (Invitrogen Life Technologies, Carlsbad, CA, USA).
  • HERV-W-env (HERV-W) and TATA-binding protein (TBP) mRNA levels were measured in triplicate samples by real-time quantitative PCR (QPCR), with SYBR green detection on iCycler iQ (Bio-Rad, Hercules, CA, USA).
  • TBP was used as a "house-keeping gene" for normalization of HERV-W mRNA expression.
  • Comparative threshold cycle (CT) was used to determine mRNA expression of HERV-W and TBP, relative to no template control.
  • the forward and reverse (5'-3') PCR primers used for HERV-W (designed from GenBank accession # AF072506) were CTTAGTGCCCCCTATGACCA (SEQ ID NO: 3) and
  • CCDS5315.1 were AACAACAGCCTGCCACCTTACG (SEQ ID NO: 5) and
  • HERV-WL Human diffuse large B cell lymphoma cells, expressing high levels of human endogenous retroviral envelope protein, were used as a source to clone endogenous retroviral protein (HERV-WL). Based on the HERV-W-env mRNA expression profile in human DLCL cells that were screened, CRL2631 DLCL has the most HERV-W mRNA expression. cDNA from CRL2631 was used to amplify HERV-env full length coding sequence. The forward and reverse primers (5 '-3') used were
  • ATGGCCCTCCCTTATCAT (SEQ ID NO: 7 )and CTAACTGCTTCCTGCTGA (SEQ ID NO: 8) (respectively).
  • Easy-A high-fidelity enzyme (Stratagene, La Jolla, CA) was used to avoid the introduction of mutation during PCR reaction.
  • PCR product was then cloned into TOPO TA vector (Life Technologies, Carlsbad, CA), clones were sequenced.
  • One clone which had the correct insert hereinafter named HERV-WL was sub cloned into expression vector pDual GC (Stratagene) for bacteria expression.
  • HERV-WL The sequence of the HERV-WL cloned from GC-derived B cell lymphoma was somewhat similar to the previously described syncytin- 1 (HERV-W) which mediates cell-cell fusions of cytotrophoblasts into syncytiotrophoblasts.
  • HERV-WL had 93% nucleotide identity compared to HERV-W.
  • HERV-WL has 2 translation frames ( Figure 6A and 6B).
  • HERV-WL translation frame 1 has The forward and reverse primers (5 '-3') used for cloning into pDual GC were AACTCTTCAATGGCCCTCCCTTATCAT (SEQ ID NO: 9) and CTAACTGCTTCCTGCTGA (SEQ ID NO: 10) (respectively).
  • the plasmid used for expression in human cells was pIRES/EGFP (Stratagene); and the forward and reverse primers (5 '-3') used for this cloning were TAGAATTC ATGGCCCTCCCTTATCAT (SEQ ID NO: 11) and AAGTCGACCTAACTGCTTCCTGCTGA (SEQ ID NO: 12) (respectively).
  • the clones of pDual GC with HERV-WL inserts were screened using the same primers used for the cloning (as stated above).
  • pDual GC vector has a coding sequence for his6 tag and a stop codon after the his6 tag at the 3' end of the insertion sequence.
  • the expression vector with the correct coding sequence for HERV-WL (now referred to as pDual GC_HERV-WL_His6) was transformed into BL21 ( DE3) cells, HERV-WL-His6 recombinant protein expression was induced by adding IPTG to the culture.
  • HERV-WL-His6 recombinant protein was purified by using Ni-NTA Superflow Columns (Qiagen Valencia, CA). The protein was dialyzed and concentrated to 2.5mg/ml.
  • This protein was used to immunize rabbits and mice to produce rabbit polyclonal and mouse monoclonal antibodies, respectively.
  • a fifteen-mer carboxyl terminal amino acid sequence (N' -TEKVKEIRDGIQRRA-C, SEQ ID NO:2) of HERV-WL was used to produce rabbit anti-peptide antibody.
  • the rabbit antibodies were purified by passing twice over pure HERV-WL affinity column.
  • the polyclonal anti-HERV-WL Abs were affinity purified, the detection of surface HERV-WL was greatly enhanced.
  • the following cell lines were detected by anti-HERV-WL Abs: CRL2630, CRL2631, RL, DB, and HT are DLCL lines; Raji and Ramos are Burkitt's lymphoma cell lines; and L540 is a Hodgkin's- derived cell line.
  • lymphoma cell lines for surface expression of HERV-WL using mouse monoclonal anti-HERV-WL antibody indicated that HERV-WL can be detected on the surface of lymphoma cells.
  • Mouse ascites fluid of anti-HERV-WL was used together with PE-conjugated goat anti-mouse F(ab')2 (as secondary antibody).
  • the use of pure expressed HERV-WL protein as a blocking agent in intracellular staining of lymphoma cells showed that the mAb is, indeed, specific for HERV-WL, see Figure 3.
  • the blocking reagent was able to reduce the level of HERV-WL in lymphoma cells to that of normal human B cell.
  • Purified IgG of anti-HERV-WL mAb was used together with PE-conjugated goat anti-mouse F(ab')2 (as secondary antibody) to detect intracellular expression of HERV-WL in DLCL lymphoma (CRL2630), compared with normal human B cells.
  • PE-conjugated goat anti-mouse F(ab')2 as secondary antibody
  • the use of pure HERV-WL protein as a blocking reagent was able to bring down the detection of HERV-WL in lymphoma cells to a level comparable to that detected in normal B cells.
  • HERV-WL expression might be detected by histology in human cancer
  • 20 archived oral squamous cell carcinoma specimens were examined by immunohistochemistry, using affinity purified rabbit polyclonal anti-HERV-WL IgG antibody.
  • the samples were deparaffinized, and the antigen was retrieved using 0.1M citrate buffer (pH 6.0). After peroxidase block, the slides coated with the samples were stained using biotinylated link followed by strepavidin-HRP, and hemnatoxylin counter stain.
  • HERV-WL was detected in mucoepidermoid carcinoma (MEC) by
  • HERV-WL was detected in ameloblastoma by immunohistochemistry by polyclonal anti-HERV-WL staining compared with normal epithelium. Ten out of ten ameloblastoma samples showed variable expression of HERV-WL in benign ameloblastoma tumor cells, with stronger expression in the tumor periphery at the interface with the connective tissue stroma.
  • Human serum was taken from 2 non-Hodgkin's lymphoma patients, 2 ovarian cancer patients, 2 breast cancer patients (hNLBCSRM), 2 cervical cancer patients (hNLCCSRM), and 2 prostate cancer patients, and human saliva from 2 non-Hodgkin's lymphoma patients and 2 ovarian cancer patients.
  • HERV-WL is increased in Ovarian Cancer, Breast Cancer and Non-Hodgkin's Lymphoma:
  • Figure 4 depicts the detection of HERV-WL protein in serum samples from normal and cancer patients.
  • the ELISA test revealed over-expression of HERV-WL in serum samples of a variety of human cancers, including no-Hodgkin's lymphoma, ovarian cancer and breast cancer.
  • Figure 5 depicts the detection of HERV-WL protein in saliva samples from normal and cancer patients.
  • Three normal saliva (hNLSLV) samples and one ovarian cancer (hOCSLV) did not have detectable HEV-WL.
  • hNHSLV2 One non-Hodgkin's Lymphoma
  • Values represent means (+SEM) of triplicate determinations.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Oncology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Hospice & Palliative Care (AREA)
  • AIDS & HIV (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention se rapporte à des polypeptides d'enveloppe de rétrovirus endogène humain (HERV-WL), à des séquences de nucléotides, à des anticorps HERV-WL. L'invention se rapporte d'autre part à des procédés de détection du cancer. L'invention se rapporte en outre à des procédés adaptés pour déterminer l'efficacité du traitement anticancer.
PCT/US2012/027289 2011-03-01 2012-03-01 Détection d'expression de rétrovirus endogène humain dans des cellules cancéreuses et des cellules saines WO2012118974A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/198,201 US20140256583A1 (en) 2011-03-01 2014-03-05 Detection of human endogenous retrovirus expression in cancer and normal cells
US15/495,627 US10370417B2 (en) 2011-03-01 2017-04-24 Methods for the diagnosis of cancer through detection of human endogenous retrovirus WL (HERV-WL) envelope expression

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161447750P 2011-03-01 2011-03-01
US61/447,750 2011-03-01

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14001304 A-371-Of-International 2012-03-01
US14/198,201 Continuation US20140256583A1 (en) 2011-03-01 2014-03-05 Detection of human endogenous retrovirus expression in cancer and normal cells

Publications (2)

Publication Number Publication Date
WO2012118974A2 true WO2012118974A2 (fr) 2012-09-07
WO2012118974A3 WO2012118974A3 (fr) 2012-10-26

Family

ID=46758496

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/027289 WO2012118974A2 (fr) 2011-03-01 2012-03-01 Détection d'expression de rétrovirus endogène humain dans des cellules cancéreuses et des cellules saines

Country Status (2)

Country Link
US (1) US20140256583A1 (fr)
WO (1) WO2012118974A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014116958A1 (fr) * 2013-01-25 2014-07-31 Regents Of The University Of Minnesota Compositions et procédés impliquant des protéines rétrovirales endogènes

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050196754A1 (en) * 2000-03-31 2005-09-08 Drmanac Radoje T. Novel nucleic acids and polypeptides
US20100074894A1 (en) * 2007-02-09 2010-03-25 Geneuro Sa Pharmaceutical composition containing antibodies directed against the herv-w envelope

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2865403B1 (fr) * 2004-01-23 2009-06-12 Biomerieux Sa Composition pour le traitement d'une pathologie associee a la msrv/herv-w

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050196754A1 (en) * 2000-03-31 2005-09-08 Drmanac Radoje T. Novel nucleic acids and polypeptides
US20100074894A1 (en) * 2007-02-09 2010-03-25 Geneuro Sa Pharmaceutical composition containing antibodies directed against the herv-w envelope

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MARSDEN ET AL.: 'Short communication: Activating stimuli enhance immunotoxin-mediated killing of HIV-infected macrophages.' AIDS RES. HUM. RETROVIRUSES. vol. 24, no. 11, November 2008, pages 1399 - 1404 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014116958A1 (fr) * 2013-01-25 2014-07-31 Regents Of The University Of Minnesota Compositions et procédés impliquant des protéines rétrovirales endogènes
US9878002B2 (en) 2013-01-25 2018-01-30 Regents Of The University Of Minnesota Compositions and methods involving endogenous retrovirus proteins

Also Published As

Publication number Publication date
WO2012118974A3 (fr) 2012-10-26
US20140256583A1 (en) 2014-09-11

Similar Documents

Publication Publication Date Title
RU2644686C2 (ru) Идентификация опухолеассоциированных антигенов для диагностики и терапии
US10202605B2 (en) Methods of identifying and treating poor-prognosis cancers
JP2007528737A (ja) Mn/caix/ca9及び腎臓癌の予後診断
EP2523687B1 (fr) Procédés pour le diagnostic et le traitement de lymphomes cutanés à cellules T ciblant le récepteur NKp46
JP7328641B2 (ja) 悪性リンパ腫又は白血病の罹患の有無の判別方法並びに白血病の治療及び/又は予防のための薬剤
KR101652962B1 (ko) 돼지열병 바이러스 생마커백신주 접종 돼지와 돼지열병 바이러스 야외주 감염 돼지의 항체 감별용 키트 및 이를 이용한 감별방법
US20230357394A1 (en) Anti-human lag-3 antibodies and their use in immunohistochemistry (ihc)
US7902347B2 (en) Human Ron-related gene variant associated with cancers
US10370417B2 (en) Methods for the diagnosis of cancer through detection of human endogenous retrovirus WL (HERV-WL) envelope expression
CN111316102A (zh) 使用甲硫氨酰-tRNA合成酶诊断胰腺癌的方法以及利用其的胰腺癌诊断试剂盒
WO2006123829A1 (fr) ANTICORPS DIRIGE CONTRE PAP2a ET SON UTILISATION DANS DES BUTS DE DIAGNOSTIC ET THERAPEUTIQUES
US20140256583A1 (en) Detection of human endogenous retrovirus expression in cancer and normal cells
KR102058150B1 (ko) 담도 세포에서 메티오닐-티알엔에이 합성효소를 이용한 담도암 진단 방법
EP2148932A1 (fr) Expression de sox11 dans des lymphomes malins
CN106957367B (zh) 抗idh1 r132h抗体及其制备方法和用途
CN112851818B (zh) 针对d-二聚体的结合蛋白及其应用和检测d-二聚体的方法
US20070264651A1 (en) Methods, compositions, and kits for the detection and monitoring of kidney cancer
JPH08500731A (ja) 診断法
US20080108070A1 (en) Methods, compositions, and kits for the detection and monitoring of colon cancer
WO2014185466A1 (fr) Procédé d'évaluation du pronostic du cancer du pancréas
JP3888695B2 (ja) ヒトlect2に対する抗体、それを産生する細胞、その測定法及び測定用キット
CN112739826A (zh) 抗体-药物缀合物的敏感性标志物
CN115028689B (zh) 特异性检测hpv6和hpv11的双特异性检测试剂盒
US10745701B2 (en) Methods of identifying and treating poor-prognosis cancers
US20040138161A1 (en) Methods of modulating proliferative conditions

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12751751

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12751751

Country of ref document: EP

Kind code of ref document: A2